Crocodilepox virus protein 157 is an independently evolved inhibitor of protein kinase R

Page created by Sheila Long
 
CONTINUE READING
Crocodilepox virus protein 157 is an independently evolved inhibitor of protein kinase R
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022                   doi:10.20944/preprints202205.0393.v1

             Crocodilepox virus protein 157 is an independently evolved inhibitor
             of protein kinase R
             M. Julhasur Rahman1,2, Loubna Tazi1, Sherry L. Haller3 and Stefan Rothenburg1*

             1
              School of Medicine, University of California Davis, Department of Medial Microbiology and
             Immunology, Davis, California, United States of America
             2
              current address: Purification Development Department, Genentech, Inc., One DNA Way, South
             San Francisco, California, United States of America
             3
             University of Texas Medical Branch at Galveston, Center for Biodefense and Emerging Infectious
             Diseases, Galveston, Texas, United States of America

             *
              Address for correspondence: Stefan Rothenburg, School of Medicine, University of California
             Davis, Department of Medial Microbiology and Immunology, Davis, CA 95616.
             rothenburg@ucdavis.edu. ORCID iD:0000-0002-2525-8230
             Keywords: poxviruses, protein kinase R, evolution, translational regulation, eIF2

                                                                                                                         1

           © 2022 by the author(s). Distributed under a Creative Commons CC BY license.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022         doi:10.20944/preprints202205.0393.v1

             Abstract

             Crocodilepox virus (CRV) belongs to the Poxviridae family and mainly infects hatchling and
             juvenile Nile crocodiles. Most poxviruses encode inhibitors of the host antiviral protein kinase R
             (PKR), which is activated by viral double-stranded (ds) RNA formed during virus replication,
             resulting in the phosphorylation of eIF2a and subsequent shutdown of general mRNA translation.
             Because CRV lacks orthologs of known poxviral PKR inhibitors, we experimentally characterized
             one candidate (CRV157), which contains a predicted dsRNA-binding domain. Bioinformatic
             analyses indicated that CRV157 evolved independently from other poxvirus PKR inhibitors.
             CRV157 bound to dsRNA, co-localized with PKR in the cytosol, and inhibited PKR from various
             species. To analyze whether CRV157 could inhibit PKR in the context of a poxvirus infection, we
             constructed recombinant vaccinia virus strains that contain either CRV157 or a mutant CRV157
             deficient in dsRNA binding in a strain that lacks PKR inhibitors. The presence of wild type
             CRV157 rescued vaccinia virus replication, while the CRV157 mutant did not. The ability of
             CRV157 to inhibit PKR correlated with virus replication and eIF2a phosphorylation. The
             independent evolution of CRV157 demonstrates that poxvirus PKR inhibitors evolved from a
             diverse set of ancestral genes in an example of convergent evolution.

                                                                                                               2
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022           doi:10.20944/preprints202205.0393.v1

             1. Introduction
             The members of the Poxviridae family are large double-stranded DNA viruses, which exclusively
             replicate in the host cytoplasm. The genomes of poxviruses range between 134 kb and 360 kb and
             encode approximately 200 genes [1]. Poxvirus members can infect vertebrates and invertebrates,
             collectively exhibiting a broad host range. Whereas some members strictly infect one or a limited
             number of host species, others can infect a large number of host species. For example, variola virus
             (VARV), the causative agent of smallpox disease, only productively infects humans. In contrast,
             other poxviruses, including cowpox viruses (CPXV), monkeypox virus (MPXV), and vaccinia
             virus (VACV) can infect a wide range of host species [2-4].
             Poxvirus infections have been reported worldwide in host species from the Crocodylia order,
             including caimans (Spectacled and Brazilian caiman) and crocodiles (Nile, saltwater, and
             freshwater crocodiles), especially in farmed caiman and crocodiles [5-9]. So far, two poxviruses
             infecting Crocodylia have been completely sequenced and assigned to the Crocodilydpoxvirus
             genus of the Chordopoxvirinae subfamily: crocodilepox virus (CRV) isolated from farmed Nile
             crocodiles [10] and saltwater crocodilepox virus (SwCRV) isolated from Australian saltwater
             crocodiles [11]. CRV mainly infects hatchling and juvenile Nile crocodiles and causes scattered,
             circular, grey to white skin lesions on the dorsal and ventral parts of the body, including the hind
             feet, mouth cavity, eyes, nostrils, head and tail regions [8,12,13]. The lesions on the eyes and head
             can impair vision and lead to skin deformities. These skin lesions can lead to further opportunistic
             infections by Dermatophilus-like bacteria or fungi. Although, in general the mortality rate is low
             in Nile crocodiles, the morbidity rate and economic impact in farmed crocodiles is high [12,14].
             Morphologically, CRV and SwCRV virions are similar to virions of orthopoxviruses, the most
             widely studied genus of Chordopoxvirinae. The virions are brick-shaped and contain a dumbbell-
             shaped central core, which is flanked by lateral bodies [6,15,16]. The genome of CRV is 190,054bp
             long and, unlike orthopoxviruses, contains a high GC content (62%). A total of 173 protein coding
             genes have been predicated in the CRV genome, which contains two short (1754 bp) inverted
             terminal repeats (ITRs). An interesting feature of CRV is that its genome lacks recognizable
             homologs of most host-immune evasion genes found in other chordopoxviruses suggesting that it
             evolved unique genes for manipulating the antiviral host response. CRV encodes 62 predicted
             proteins that don’t show apparent sequence homology with non-Crocodylidpoxvirus proteins.
             Forty-eight of these protein encoding genes are located in the terminal regions [10].
             The ability of poxviruses to enter host cells is independent of species-specific cellular receptors,
             which allows poxviruses to be able to enter into many different cell types [2,17]. The successful
             replication of poxviruses depends on their ability to subvert host immune responses [2,3]. During
             virus infections, host cells induce type I interferons (IFNs) and other antiviral host pathways,
             leading to the establishment of an antiviral state [18]. Poxviruses accommodate a variety of
             different immunomodulatory genes, which interfere with host antiviral proteins to allow virus
             replication [19]. One of the IFN-induced host antiviral proteins is protein kinase R (PKR), which
             is targeted by many viruses, including poxviruses [20-24]. PKR is present at intermediate levels in
             unstimulated cells as an inactive monomer. Upon detection and binding of double-stranded (ds)
             RNA produced during virus replication and transcription, PKR undergoes conformational changes,
             which lead to PKR activation via homodimerization and autophosphorylation. Activated PKR
             phosphorylates the alpha subunit of eukaryotic translation initiation factor 2 (eIF2), which leads
             to the suppression of general mRNA translation and in turn restricts virus replication [25-32]. In
             addition, PKR can also inhibit virus replication by inducing proinflammatory responses and cell
             death in infected cells [33-36]. Most mammalian poxviruses inhibit PKR by encoding orthologs of

                                                                                                                 3
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022         doi:10.20944/preprints202205.0393.v1

             two vaccinia virus PKR inhibitors: E3 and K3 [37,38]. E3 inhibits PKR activation by binding to
             dsRNA and inhibiting PKR homodimerization [22,39-41]. K3 is a pseudosubstrate inhibitor of PKR,
             which restricts eIF2a and PKR interactions [21,23,42].
             CRV lacks homologs of known PKR inhibitors [10]. It is therefore unknown how it inhibits the
             PKR pathway. To investigate this, we performed a BLAST database searches and identified a
             putative viral PKR inhibitor from open reading frame (ORF) 157 of the CRV genome, CRV157.
             CRV157 was previously characterized in silico as a putative dsRNA binding protein [43]. CRV157
             contains a putative dsRNA binding domain (dsRBD) and an uncharacterized C-terminal domain
             that does not show homology to other known proteins. In this study, we investigated the hypothesis
             that CRV157, with its putative dsRNA binding domain, might act as a PKR antagonist. The results
             shown here indicate that CRV157 can bind dsRNA, inhibit PKRs from different species, and that
             its dsRNA binding ability is essential for PKR inhibition. Furthermore, CRV157 was able to rescue
             replication of a recombinant VACV that lacks both PKR inhibitors E3L and K3L. These results
             indicate that CRV is an independently evolved PKR inhibitor.

             2. Materials and Methods

             2.1 Cell lines
             European rabbit RK13 (kindly provided by Dr. Bernard Moss), HeLa (ATCC#CCL-22), HeLa-
             PKRkd (knockdown) (kindly provided by Dr. Charles Samuel [35], and RK13+E3+K3 (RK13 cells
             stably express VACV E3L and K3L) [44] cell lines were maintained in Dulbecco’s Modified
             Essential Medium (DMEM, Life Technologies) supplemented with 5% fetal bovine serum (FBS,
             Fischer Scientific) and 25µg/ml gentamycin (Quality Biologicals). RK13+E3+K3 cells were
             additionally supplemented with 500µg/ml geneticin (Life Technologies) and 300µg/ml zeocin
             (Life Technologies). All cells were incubated at 37° C, 5% CO2.

             2.2 Plasmids
             ORFs of all PKRs from the indicated species and viral genes were cloned under the control of a
             SV40 promoter into the pSG5 vector (Stratagene) for transient expression. Construction of
             knockdown resistant human PKR, mouse PKR, European rabbit PKR, and Chinese hamster PKR
             was previously reported [45,46]. Chicken PKR (identical to XM_015283611.2) gene was cloned
             from a 14-day old chicken embryo (Gallus gallus). Armenian hamster PKR and pig PKR were
             cloned from AHL-1 cells (ATCC-CCL-195) and swine PK15 cells (ATCC-CCL-33), respectively.
             CRV157 (GeneID: 4363410) was obtained from gBlocks®Gene fragments (IDT) and cloned into
             the TOPO-TA vector (Invitrogen). The CRV157 ORF was then subcloned into the pSG5 vector
             with SacI and XhoI restriction sites. CRV157-K48A/K49A, CRV157 delC, and CRV157 delN
             plasmids were generated by site-directed mutagenesis or deletion mutation PCR using pfu high-
             fidelity DNA polymerase (Invitrogen). For site-directed mutagenesis, PCR was performed using
             pSG5-CRV157 as a template with the primers containing lysine to alanine mutations (AAA to
             GCA and AAG to GCG) from nucleotide position 142 to 146). For the C-terminal deletion,
             nucleotide position 1 to 198 and N-terminal deletion, nucleotide position 199 to 384 were PCR
             amplified and cloned into pSG5 vector. To make flag epitope-tags at the C-termini of VACV E3L,
             CRV157, CRV157-K48A/K49A, CRV157 del C, and CRV157 del N plasmids, these ORFs were
             cloned into a pSG5 vector containing two in-frame flag tag sequences. CRV157-mCherry and

                                                                                                               4
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022          doi:10.20944/preprints202205.0393.v1

             chicken PKR-EGFP fusion genes were constructed by fusion PCR, and constructs were cloned
             into the pSG5 vector. All plasmid constructs were sequenced to confirm the correct sequences.

             2.3 Transfection and luciferase-based reporter (LBR) assays
             Luciferase assays were performed following the protocol described previously [45]. Briefly, HeLa-
             PKRkd cells (5X104 cells/ well) were seeded in 24 well plates a day before the transfection. For
             each transfection reaction mix, 0.05µg of firefly luciferase (Promega) and indicated PKR (0.2µg)
             plasmids were co-transfected with 0.8µg of either E3L, CRV157, CRV157-K48A/K49A, CRV157
             del C, or CRV157 del N plasmids using GenJet (Signagen) with transfection reagent to DNA ratios
             of 2:1. For titration experiments, indicated concentrations of CRV157 or CRV157 del C plasmids
             were co-transfected. Each transfection was done in triplicate, and plasmid DNA concentration for
             each transfection mix was kept constant by adding an appropriate amount of pSG5 vector plasmid
             where necessary. After 48 h, whole cell lysates were extracted using mammalian lysis buffer
             (Goldbio) followed by the addition of luciferin (Promega). Luciferase activity was determined in
             a Glowmax luminometer (Promega). Each experiment was conducted at least three times, and
             representative experiments are shown.

             2.4 Viruses and infection assays
             Vaccinia Virus Copenhagen strain, VC-2, was used as a parental strain to make all the recombinant
             viruses used in this study. VC-2 and its derivative vP872 (∆K3L) [21] were kindly provided by Dr.
             Bertram Jacobs. The construction of the vP872 derivative VC-R4 (∆E3L, ∆K3L, in which E3L
             was replaced by EGFP) was described previously [47]. Recombinant viruses including VC-R4-
             VACV E3L, VC-R4-CRV157 and VC-R4-CRV157-K48A/K49A were generated into the VC-R4
             backbone by scarless integration of the indicated genes into the E3L locus of VC-R4 as described
             [47]. All the recombinant viruses were plaque purified three times. PCR amplification and
             sequencing of recombinant virus integration sites were done to confirm successful integration and
             correct sequence of CRV157, VC-R4-CRV157-K48A/K49A, or E3L.
             Replication kinetics of VC-R4 recombinants were tested in the RK13 cell line. Briefly, RK13 cells
             (6 X 105 cells) were plated in 6-well plates a day before the infection. RK13 cells were infected at
             a MOI of 0.01 with VC-R4, or VC-R4 derived recombinant viruses. Virus samples were sonicated
             before the infection (2 times for 15s) and diluted in 2.5% FBS supplemented DMEM media. Each
             infection at each different time point was done in duplicate. Growth medium from each well was
             aspirated to add virus inocula to respective wells and then incubated at 37˚C for 1 hour. After 1-
             hour incubation period, virus inocula were removed, and each well was washed twice with
             phosphate buffered saline (PBS), and fresh DMEM media supplemented with 5% FBS was added.
             Cells were then scraped and collected along with the media at 0, 12, 24, 48, 72, and 96 hpi.
             Collected viruses were then subjected to three rounds of freeze/thaw cycles followed by sonication
             (2 times for 30s, 50% amplitude) in a cup sonicator (Qsonica Q500). Titration of collected viruses
             was done in RK13+E3+K3 cells by measuring plaque forming units per ml (pfu/ml).

             2.5 Poly I:C Pull-down assays
             Pull-down assays were performed following the protocol previously described [48]. Sepharose
             beads were prepared by dissolving one gram of lyophilized CNBr-activated sepharose powder
             beads (17-0430-01; GE Healthcare Life Sciences) in 40 mL of 1mM HCl followed by mixing 10
             times by end-to-end rotation. The beads were then pelleted by centrifuging at 1000 X g for 5
             minutes; the supernatant was then removed without disturbing the beads. The addition of 1mM

                                                                                                                5
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022          doi:10.20944/preprints202205.0393.v1

             HCl and removal of supernatant after centrifugation was repeated 4 times. After the final wash,
             HCl was removed, and the beads were ready for Poly I:C conjugation. A stock solution of poly I:C
             was prepared in coupling buffer (0.1 M NaHCO3, 0.5M NaCl, pH 8.3) at 10mg/mL concentration.
             One gram of prepared sepharose beads was mixed with 2mL of poly I:C stock solution in a total
             volume of 4mL in coupling buffer and mixed by end over end rotation at RT for 2 hours.
             Unconjugated poly I:C was removed by washing the beads 5 times with equal volumes of coupling
             buffer and centrifugation at 1000 X g for 5 minutes between washes and removal of the
             supernatant. Beads were then incubated with 0.1M Tris HCl, pH 8 for 2 hours at RT to block the
             unconjugated active sites on the beads followed by 3 cycles of washing of conjugated beads with
             alternating low-pH (0.1Msodium acetate, 0.5M NaCl, pH 4.0) and high-pH (0.1MTris-HCl,
             0.5MNaCl, pH 8.0) buffer (5 volumes of bead volume of each pH buffer were used). After the
             final wash, beads were stored in Co-IP lysis buffer (20mMTris-HCl, pH 8, 137 mM NaCl, 10%
             glycerol, 1% Triton-X, 2 mM EDTA) before use as a 50% bead slurry (equal volume of buffer
             added to the beads).
             HeLa-PKRkd cells were transfected at 70%-80% confluency in 6-well plates with 3µg of plasmids
             pSG5-flag, pSG5-E3L-flag, pSG5-CRV157-flag or pSG5-CRV157-K48A/K49A-flag. After 48-
             hours, the media was removed and washed with PBS buffer followed by cell lysis with 300 µl of
             lysis buffer (20mM Tris-HCl, pH 8, 137 mM NaCl, 10% glycerol, 1% Triton X-100, 2 mM EDTA,
             protease inhibitor) on ice. For the pre-pull-down assay, 30µl of lysed samples were separated, and
             the rest of the samples were centrifuged at 5400 X g for 10 minutes to remove cellular debris. Pull-
             down of poly I:C was done by adding 50µl of poly I:C bead slurry to each cell lysate sample
             followed by end over end rotation at RT for 2 hours and subsequent centrifugation at 5400 X g for
             2 minutes and removal of the supernatant. The samples were then washed 3 times with 200 µl of
             lysis buffer, and after the final wash, proteins were released from the poly I:C beads, samples were
             resuspended in 120 µl of Laemmli buffer (6X SDS, β-mercaptoethanol) and denatured at 95˚C for
             10 minutes. The samples were then centrifuged at 5400 X g for 2 min, and supernatants were
             collected and subjected to Western blotting.

             2.6 Western blot
             Protein lysates from HeLa-PKRkd cells transfected with the indicated plasmid constructs (3µg)
             were collected 48 hours post transfection in 1% sodium dodecyl-sulfate (SDS) in PBS (VWR).
             Protein lysates from RK13 cells infected with either VC-R4 (∆E3L∆K3L), VC-2, VC-R4-VACV
             E3L, VC-R4-CRV157, or VCR2-CRV157-K48A/K49A at MOI= 3 were collected at 6 hours post
             infection (hpi) in 1% SDS. All protein lysates were sonicated (twice for 5s at 50% amplitude) to
             shear genomic DNA, separated by 12% SDS- polyacrylamide gel electrophoresis and blotted onto
             Hybond-PVDF (GE Healthcare) membranes using a methanol-based wet transfer apparatus
             (BioRad). Membranes were blocked for 1 hr at room temperature with either SuperBlock blocking
             buffer (ThermoFisher) when detecting flag and ß-actin, 5% (wt/vol), BSA when detecting
             phosphorylated eIF2a, or 5% (wt/vol) nonfat milk when detecting total eIF2a in TBST buffer (20
             mM Tris, 150 mM NaCl, and 0.1% Tween-20 pH 7.4). The blots were then incubated overnight at
             4˚C with primary antibody either mouse anti-D (1:1000, FLAG, abm), rabbit eIF2a-P (1:1000,
             Santa Cruz Biotechnology, sc-101670), rabbit total eIF2a (1:1000, Santa Cruz Biotechnology, sc-
             11386) or mouse anti-b-actin (1:1000, Sigma-Aldrich) diluted in TBST buffer. The membranes
             were then washed three times for 5 minutes each in 1X TBST buffer followed by incubation with
             horseradish peroxidase conjugated secondary antibodies (donkey anti-rabbit-HRP or donkey anti-
             mouse-HRP, Life Technologies) in blocking buffer for 1 h at room temperature. The membranes

                                                                                                                6
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022          doi:10.20944/preprints202205.0393.v1

             were then washed 3 times 10 minutes each in TBST buffer. After washing the membranes, proteins
             bound by the secondary antibody were detected with Proto-Glo ECL (National Diagnostics), and
             images were taken with either a Kodak-4000MM Image Station or iBright (Invitrogen).

             2.7 Phylogenetic analysis
             The dsRBDs protein sequences from 62 viral proteins were aligned using MUSCLE [49]. A
             maximum likelihood phylogenetic tree was then generated from the multiple sequence alignment
             using PhyML 3.0 with 100 bootstrap replicates [50]. The resulting tree was visualized using the
             program FigTree v1.3.1 (http://tree.bio.ed.ac.uk/software/figtree/).

             2.8 Confocal microscopy
             To determine the localization of PKR and CRV-157, HeLa-PKRkd cells were plated either on glass
             coverslips in 12 well plates or on 35-mm coverglass bottom dishes (MatTek). At 40-50%
             confluency, the cells were co-transfected with EGFP-tagged chicken PKR (0.5µg), and mCherry
             tagged CRV157 (0.5µg) and incubated at 37˚C for 48h. After the incubation, the media was
             removed, and cells were washed with PBS. The cells were then fixed with 2% formaldehyde and
             incubated for 20-30 min, followed by PBS washing to remove formaldehyde. Following
             formaldehyde removal, cells were incubated with 50mM glycine for 5-10 min at RT and then
             washed with PBS. The cells were then incubated with nuclear stain, DAPI (300nM), for 15-20 min
             and washed (x3) with PBS. Glass coverslips were removed and placed upside down on glass slides
             with mounting media. Excess mounting media were absorbed from the edges using filter paper
             and edges were sealed by applying nail polish. After nuclear staining, the cells on the glass bottom
             dish were kept in PBS.
             Samples were then observed with an inverted Carl Zeiss LSM 880 confocal microscope using
             excitation wavelengths of 405nm (DAPI), 514nm (EGFP), and 633nm (mCherry) with 40x EC
             Plan-Neofluar oil objective with a 1.3 numerical aperture (NA). Fluorescence was detected using
             a gallium arsenide phosphide (GaAsP) detector to ensure improved signal to noise ratio.
             Fluorescent images were obtained by sequential scanning of each channel to ensure reliable
             quantitation of colocalization. Images of random fields were acquired, and channels were merged
             in Zeiss Zen microscope software during acquisition, which allowed qualitative judgments about
             colocalization of CRV157-mCherry and chicken PKR-EGFP signals. Confocal images were then
             imported to Fiji (v2.00-rc-68/1.52f) [51] software for colocalization analysis.

             2.9 Quantitative colocalization analysis (QCA)
             Images acquired from the confocal microscope were processed using Fiji and the JACoP plugin
             [51,52]. Briefly, the background of channels with mCherry and EGFP were subtracted from the
             control confocal images (no fluorescent/fluorescence) by using the image calculator tool.
             Furthermore, background was corrected using Coste’s automatic threshold for all channels.
             Pearson’s correlation coefficients (PCC) were calculated using Pearson’s coefficient tab of the
             JACoP plugin. PCC values range from -1.0 to 1.0, where -1.0 implies no overlap and 1.0 indicates
             complete colocalization [53]. The data obtained were analyzed in Excel.

                                                                                                                7
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022        doi:10.20944/preprints202205.0393.v1

             3. Results

             3.1 CRV157 contains a dsRNA-binding domain
             In order to identify putative PKR inhibitors in other poxviruses, we performed PSI-BLAST
             searches of poxvirus sequences using default parameters with homologs of E3 and K3 from various
             poxviruses. For example, when we used the dsRNA binding domain of VACV E3 or the racoonpox
             virus E3 homolog as bait, CRV157, a putative protein encoded by CRV was identified in iteration
             2 but stayed below the PSI-BLAST threshold of 0.005 through iteration 4, when no new sequences
             were identified above the threshold. This indicated that CRV157 might be a dsRNA binding
             protein, distantly related to E3 orthologs. CRV157 was previously identified as a putative dsRNA
             binding protein when CRV proteins of unknown functions were searched against the NCBI
             database using PSI-BLAST [43]. The authors concluded that CRV157 was not an ortholog of the
             E3 family. To follow up, we extended our search for E3 homologs to include all viral proteins in
             the database. While dsRBDs from dozens of non-poxviruses were detected after iterations 2 and
             3, CRV157 was only detected after iteration 4 and its ortholog (ORF198) from saltwater
             crocodilepox virus (SwCRV) only after iteration 5. To study the relationship among viral dsRBDs,
             we performed a phylogenetic analysis from a multiple sequence alignment containing dsRBDs
             from various viruses. In this analysis, CRV157 and SwCRV198 were found in a different clade
             than E3 sequences from other poxviruses (Fig. 1). Even though the basal branches were not
             supported by high bootstrap values (a notable problem for phylogenetic analyses of dsRNA-
             binding proteins because only a few amino acids are conserved [54]) these analyses, together with
             the PSI-BLAST searches further support the notion that CRV157 does not share an immediate
             ancestor with the poxvirus E3 family.
             When PSI-BLAST searches with the full CRV157 sequences of the whole NR database were
             performed with initial BLAST thresholds set to 1, various dsRBDs of ribonuclease III proteins
             from Streptomyces species were discovered during the second iteration. The detected homology
             was not restricted to the dsRBD but extended a further 45 amino acids into the C-terminus (Fig.
             2A). A multiple sequence alignment with other dsRBDs including VACV E3 homologs myxoma
             virus 029 and sheeppox virus 34, showed a deletion between beta sheets 1 and 2 in CRV157, as
             previously reported [43]. Sequence identities calculated from the alignment including only the
             dsRBDs showed highest sequence identity of CRV157 and SwCRV198 with S. gossypiisoli
             ribonuclease III (Fig. 2B). Combined, these data indicate that CRV157 is not an ortholog of the
             poxvirus E3 family, but originated independently.

             3.2 CRV157 is a dsRNA binding protein.

             Because CRV157 contains a deletion between beta sheets 1 and 2, also called region 2, which has
             been shown to be important for dsRNA binding in other dsRBDs, we experimentally tested its
             dsRNA-binding capability in a pull-down assay using the synthetic dsRNA analog
             polyinosinic:polycytidylic acid (poly I:C). We assessed poly I:C pull-down of CRV157 and a
             mutant of CRV157 (CRV157-K48A/K49A) in which key lysine residues that were shown to be
             essential for dsRNA binding in other dsRBD have been replaced by alanine residues [40]{Bycroft,
             1995 #51}[48,55,56]. In order to be able to detect and to pull-down the proteins, we added C-
             terminal FLAG epitope tags to CRV157, CRV157-K48A/K49A, and E3L in the mammalian
             expression vector pSG5. For the pull-down assay, HeLa-PKRkdcells were transiently transfected

                                                                                                              8
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022           doi:10.20944/preprints202205.0393.v1

             with either pSG5 vector, VACV E3L, CRV157, or CRV157-K48A/K49A. Cell lysates from these
             cells were incubated with poly I:C-conjugated sepharose beads and bound proteins were subjected
             to Western blot analysis. While all three proteins were detected in the initial lysates, only E3 and
             CRV157 were detected in the pull-downs, but not CRV157-K48A/K49A (Fig. 3). These results
             demonstrate that CRV157 is a dsRNA binding protein.

             3.3 CRV157 is a PKR inhibitor

             To test whether CRV157 can inhibit PKR, we used an established luciferase-based reporter (LBR)
             assay [45]. In this assay, PKR, potential PKR inhibitors and a luciferase reporter are co-transfected
             into PKR-depleted or PKR-deficient cells and luciferase activity is used as a proxy for translation,
             where low luciferase activities indicate high PKR activity, whereas high luciferase activities
             indicate low PKR activity. In this assay PKR is activated during transient transfection, likely by
             dsRNA formed from overlapping RNA transcripts of the transfected plasmids [57]. We co-
             transfected either empty vector, mouse or chicken PKR with empty vector, E3L, CRV157 or
             CRV157-K48A/K49A. Co-transfection of both E3L and CRV157 resulted in increased luciferase
             activities, which is indicative of PKR inhibition, whereas CRV157-K48A/K49A had no effect
             (Fig. 4A). To analyze the contribution of the two CRV157 domains to PKR inhibition, we
             generated FLAG-tagged constructs encoding either the N-terminal (dsRBD) or C-terminal
             domains (Fig. 4B) and performed LBR assays with these and full-length constructs. The construct
             containing the dsRBD resulted in comparable luciferase activity as full-length CRV157, whereas
             the C-terminal domain did not affect PKR activity (Fig. 4C). Western blots of transfected cells
             showed comparable expression of CRV157 and the N-terminal domain, whereas no expression
             was observed for the C-terminal domain (Fig. 4D). Co-transfection of increasing amounts of
             CRV157 and the CRV157 N-terminus with chicken PKR revealed that full-length CRV157
             inhibited PKR more efficiently than the dsRBD alone (Fig. 4E).

             3.4 CRV157 can inhibit PKR from multiple species

             Multiple studies have previously shown that PKR inhibitors from poxviruses can show species
             specific PKR inhibition [45,46,58-60]. To investigate whether CRV157 exhibits species-specific
             PKR inhibition, we performed the LBR assay with CRV157 or VACV E3L as control, and PKR
             from the following seven vertebrate species: human, mouse, European rabbit, pig, Armenian
             hamster, Chinese hamster or chicken (Fig. 4.5). The PKR gene from the Nile crocodile, the natural
             CRV host, has not yet been sequenced and was therefore unavailable for these experiments. All
             tested PKRs were inhibited by E3 and CRV157, although to different extents. Human and mouse
             PKR were only weakly to moderately inhibited, whereas chicken PKR was most strongly inhibited.
             In all cases, E3 showed stronger inhibition than CRV157.

             3.5 Colocalization of CRV157 with PKR

             To investigate whether CRV157 colocalizes with PKR, we fluorescently labelled CRV157 and
             chicken PKR at the C-termini, with mCherry (CRV157-mCherry) and EGFP (chicken PKR-
             EGFP), respectively. We co-transfected HeLa-PKRkd cells with CRV157-mCherry and chicken

                                                                                                                 9
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022          doi:10.20944/preprints202205.0393.v1

             PKR-EGFP to analyze their localization in cells using confocal microscopy. Confocal images were
             taken for quantitative colocalization analysis (QCA), which measures the degree of spatial
             coincidence between CRV157-mCherry and chicken PKR-EGFP using Fiji ImageJ analysis
             software. The degree of co-localization of CRV157 and chicken PKR was calculated from at least
             15 representative confocal images from three independent experiments and is shown as the average
             of the Pearson’s correlation coefficients (PCC). The PCC value (0.77) from QCA indicates a strong
             correlation for the colocalization of CRV157 and chicken PKR in the cell cytoplasm (Fig. 6A) [61].
             Representative images of the localizations are shown (Fig. 6B).

             3.6 CRV157 can rescue the replication of an attenuated VACV strain that lacks its PKR
             inhibitors

             The best test to analyze if CRV157 is a biologically meaningful PKR inhibitor, is to insert it into
             a VACV strain that lacks its PKR inhibitors and test whether it can rescue replication. We therefore
             inserted either CRV157, CRV157-K48A/K49A, or VACV E3L (as positive control) into the
             replication incompetent VACV strain VC-R4, which lacks both E3L and K3L. All the transgenes
             were integrated into VC-R4 by a method that allows seamless integration of transgenes into the
             E3L locus and is driven by the same endogenous E3L promoter [47]. To compare the replication
             efficiency of these viruses relative to VC-2 (parental to all viruses tested here), vP872 (∆K3L),
             and the parental VC-R4 (∆E3L∆K3L), we infected rabbit kidney RK13 cells at MOI= 0.01 with
             these viruses over the course of 96 hours (Fig. 7A). Cell lysates were collected at different time
             points and viruses were titered on the permissive RK13+E3+K3 cell line. The replication kinetics
             indicate that VC-R4 and VC-R4+CRV157-KK-AA were replication defective. VC-R4 containing
             E3L (VC-R4+E3L) replicated comparably well as VC-2 and vP872 at all time points, which
             reached plateaus after 48 hours. VC-R4 containing CRV157 (VC-R4+CRV157) showed
             replication, but it lagged behind the replication of the E3L-containing viruses at 12, 24 and 48
             hours by about 10-fold. However, replication was comparable at 72 and 96 hours to the E3L-
             containing viruses. VC-R4+CRV157 replicated to more than 4 orders of magnitude higher titers
             than VC-R4 and VC-R4:CRV157-KK-AA viruses after 72 and 96 hours.
             Since eIF2a phosphorylation more directly demonstrates the effects of PKR inhibitors, we also
             monitored eIF2a phosphorylation levels in virus-infected RK13 cells by Western blot analysis
             (Fig. 7B). To analyze eIF2a phosphorylation, we infected RK13 cells with the above mentioned
             VACV strains at MOI=3 for 6 hours. In RK13 cells, VC-R4+CRV157-K48A/K49A infection
             induced high levels of eIF2a phosphorylation, which was comparable to that that seen in VC-R4
             infected cells. Whereas, VC-2, vP872 and VC-R4+E3L infections led to low eIF2a
             phosphorylation levels, intermediate eIF2a phosphorylation levels were observed in VC-
             R4+CRV157 infected cells. Thus, PKR sensitivity and virus replication correlated well with eIF2a
             phosphorylation levels in infected RK13 cells.

             4. Discussion

             Poxviruses infecting crocodilians pose threats for both farm-raised and wild populations [12,14,16].
             To date, the full genome sequences from two such poxviruses (CRV and SwCRV) have been
             described, which form a monophyletic clade and have been assigned to the crocodylidpoxvirus

                                                                                                               10
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022           doi:10.20944/preprints202205.0393.v1

             genus [10,11]. In depth genomic characterization of multiple SwCRV isolates, showed strong
             evidence of recombination within the SwCRV clade [62]. To the best of our knowledge, however,
             no functional characterizations of any crocodylidpoxvirus proteins have been reported. Here we
             show that CRV157 is a PKR inhibitor and highlight the importance of the role of the PKR pathway
             in crocodylidpoxvirus infections.
             Together with an earlier study [43], all evidence presented here indicates that CRV157 evolved
             independently from the other poxvirus PKR inhibitors. While it is hard to trace the evolutionary
             origins of individual dsRBDs because of the rapid evolution of the dsRBD having only a few
             conserved amino acid residues [54], the extended homology in the C-terminus of CRV157 with
             Streptomyces sp. RNase III indicates that it might have been horizontally transferred from an
             unsampled bacterium. However, the currently available sequences do not allow us to infer the
             origin of CRV157 with high confidence. While most poxvirus proteins show higher sequence
             identities with eukaryotic proteins in bioinformatic analyses, some poxvirus proteins appear to be
             more similar to bacterial proteins indicating possible transfer events between bacteria and viruses
             [63]. Indeed, several CRV proteins show higher sequence identities to bacterial proteins than to
             eukaryotic proteins [10]. One of these examples is CRV051, which shows highest sequnce
             identities (about 32%) with prokaryotic serine recombinases [64].
             Although CRV157 evidently contains a dsRBD as previously reported [43], it was important to
             determine, if it was actually functional, especially in light of a deletion between beta sheets 1 and
             2 (region 2), which includes amino acid residues that have been shown to be involved in dsRNA
             interactions in other dsRBDs [65]. Poly (I:C) pull-down of wild type CRV157, but not of CRV157-
             K48A/K49A, showed that CRV157 is a dsRNA binding protein. This binding activity correlated
             with the inhibition of PKR, which was only inhibited by wild type CRV157 but not CRV157-
             K48A/K49A. Importantly, CRV157, but not CRV157-K48A/K49A, was able to rescue replication
             of VACV missing K3L and E3L genes in RK13 cells. The finding that titers of the CRV157-
             containing virus were about 10-fold lower out to 3 days of infection than of the E3L-containing
             viruses, however, indicated that CRV157 inhibited rabbit PKR sub-optimally, at least in the
             context of VACV infection. This correlated with the intermediate eIF2a phosphorylation levels
             induced by VC-R4+CRV157, and the findings that CRV157 was not as potent in inhibiting PKR
             from all tested vertebrates as VACV E3 in the LBR assay. It is possible that PKR from the natural
             host, the Nile crocodile, might be more sensitive to CRV157 inhibition, which could not be tested
             because the PKR gene from this species is not known, or that it works sub-optimally in the context
             of VACV infection, or in the tested mammalian cells. It is also possible that the intermediate PKR
             inhibition might be associated with the longevity of the CRV infection in Nile crocodiles, which
             can often persist for many months [6,12]. Another possibility is that intermediate PKR inhibition
             may have unexpected consequences such as the functional activation of the pro-inflammatory NF-
             kB pathway, as recently shown [36]. In this case, limited inhibition of translation resulted in NF-
             kB activation and the translation of induced mRNA, whereas no PKR inhibition abolished
             translation of the induced mRNAs.
             An interesting observation from the LBR assay was that the individual tested PKRs appeared to
             exhibit differential sensitivity to both CRV157 and E3, with human PKR being inhibited least and
             chicken PKR being inhibited strongest, indicating that some PKRs might be more sensitive to viral
             dsRNA binding proteins than others. Notably, expression of the dsRBD of CRV157 alone
             (CRV157 del C) did not inhibit PKR as efficiently as full length CRV157, indicating that the
             uncharacterized C-terminal domain of CRV157 might be also involved in PKR inhibition. This is
             reminiscent of the situation found in VACV E3 and the variola virus E3 homolog, for which the

                                                                                                                11
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022            doi:10.20944/preprints202205.0393.v1

             N-terminal Za domain was shown to be important for PKR inhibition in addition to the C-terminal
             dsRBD in yeast assays [41,66]. In the context of vaccinia virus infection, an N-terminally truncated
             E3L gene resulted in altered eIF2a phosphorylation levels at 6 hours, but led to increased eIF2a
             phosphorylation at 9 and 12 hours after infection [67]. These findings show that PKR inhibition
             can be influenced by domains adjacent to the dsRBD.

             5. Conclusions
             Poxviruses evolved multiple inhibitors of antiviral host proteins, often targeting various steps in
             antiviral pathways [19]. The critical role of PKR in inhibiting virus replication is highlighted by
             the fact that most mammalian poxviruses contain two inhibitors, named E3 and K3 in vaccinia
             virus, which either prevent PKR activation, or PKR interaction with its substrate IF2a. Notably,
             E3 and K3 orthologs are missing in avipoxviruses and crocodylidpoxviruses [37,38].
             Avipoxviruses, possess a protein with homology to the eIF2a-binding and protein phosphatase 1
             (PP1)-binding region of the DNA damage-inducible protein 34 (GADD34). Similar to GADD34,
             canarypox virus protein 231, was able to suppress PKR-toxicity in a yeast assay, in which it also
             promoted PKR-mediated eIF2a dephosphorylation [68]. The data presented here show that
             CRV157 is a fourth PKR inhibitor found in poxviruses and thus provides a compelling example
             for convergent evolution.
             Because of the negative impact of crocodilepox viruses on crocodile farming, an important goal
             would be to develop an animal vaccine for this industry. Because vaccinia virus with targeted
             deletion in the PKR inhibitor E3L has been shown to protect mice and rabbits from lethal
             challenges with ectromelia virus and rabbitpox virus, respectively [69,70], CRV or SwCRV strains
             with deletions in CRV157 or SwCRV198 might be promising candidates for such vaccines.

             Acknowledgments:

             Author Contributions: Conceptualization, M.J.R. and S.R.; methodology, M.J.R., L.T., S.H. and
             S.R.; validation, M.J.R.; formal analysis, M.J.R., L.T., and S.R.; investigation, M.J.R., L.T., S.H.
             and S.R.; resources, M.J.R., L.T., S.H. and S.R. and T.S.; data curation, M.J.R., L.T., and S.R.;
             writing— original draft preparation, M.J.R. and S.R.; writing and editing, M.J.R., L.T., S.H. and
             S.R.; visualization, M.J.R., L.T., and S.R.; supervision, S.R.; project administration, S.R.; funding
             acquisition, S.R. All authors have read and agreed to the published version of the manuscript.

             Funding: This research was partially funded by grant AI114851 (to S.R.) from the National
             Institute of Allergy and Infectious Diseases, National Institutes of Health, and start-up funds from
             the University of California, Davis (to S.R.).

             Conflicts of Interest: All authors declare no conflict of interest.

                                                                                                                 12
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022            doi:10.20944/preprints202205.0393.v1

             References

             1.      Hendrickson, R.C.; Wang, C.; Hatcher, E.L.; Lefkowitz, E.J. Orthopoxvirus genome
                     evolution: the role of gene loss. Viruses 2010, 2, 1933-1967, doi:10.3390/v2091933.
             2.      McFadden, G. Poxvirus tropism. Nat Rev Microbiol 2005, 3, 201-213,
                     doi:10.1038/nrmicro1099.
             3.      Haller, S.L.; Peng, C.; McFadden, G.; Rothenburg, S. Poxviruses and the evolution of host
                     range and virulence. Infect Genet Evol 2014, 21, 15-40,
                     doi:10.1016/j.meegid.2013.10.014.
             4.      Silva, N.I.O.; de Oliveira, J.S.; Kroon, E.G.; Trindade, G.S.; Drumond, B.P. Here, There, and
                     Everywhere: The Wide Host Range and Geographic Distribution of Zoonotic
                     Orthopoxviruses. Viruses 2020, 13, doi:10.3390/v13010043.
             5.      Jacobson, E.R.; Popp, J.A.; Shields, R.P.; Gaskin, J.M. Poxlike skin lesions in captive
                     caimans. J Am Vet Med Assoc 1979, 175, 937-940.
             6.      Pandey, G.S.; Inoue, N.; Ohshima, K.; Okada, K.; Chihaya, Y.; Fujimoto, Y. Poxvirus
                     infection in Nile crocodiles (Crocodylus niloticus). Res Vet Sci 1990, 49, 171-176.
             7.      Penrith, M.L.; Nesbit, J.W.; Huchzermeyer, F.W. Pox virus infection in captive juvenile
                     caimans (Caiman crocodilus fuscus) in South Africa. J S Afr Vet Assoc 1991, 62, 137-139.
             8.      Huchzermeyer, F.W.; Wallace, D.B.; Putterill, J.F.; Gerdes, G.H. Identification and partial
                     sequencing of a crocodile poxvirus associated with deeply penetrating skin lesions in
                     farmed Nile crocodiles, Crocodylus niloticus. Onderstepoort J Vet Res 2009, 76, 311-316,
                     doi:10.4102/ojvr.v76i3.35.
             9.      Marschang, R.E. Viruses infecting reptiles. Viruses 2011, 3, 2087-2126,
                     doi:10.3390/v3112087.
             10.     Afonso, C.L.; Tulman, E.R.; Delhon, G.; Lu, Z.; Viljoen, G.J.; Wallace, D.B.; Kutish, G.F.;
                     Rock, D.L. Genome of crocodilepox virus. J Virol 2006, 80, 4978-4991,
                     doi:10.1128/JVI.80.10.4978-4991.2006.
             11.     Sarker, S.; Isberg, S.R.; Milic, N.L.; Lock, P.; Helbig, K.J. Molecular characterization of the
                     first saltwater crocodilepox virus genome sequences from the world's largest living
                     member of the Crocodylia. Sci Rep 2018, 8, 5623, doi:10.1038/s41598-018-23955-6.
             12.     Huchzermeyer, F.W.; Huchzermeyer, K.D.; Putterill, J.F. Observations on a field outbreak
                     of pox virus infection in young Nile crocodiles (Crocodylus niloticus). J S Afr Vet Assoc
                     1991, 62, 27-29.
             13.     Huchzermeyer, F.W. Diseases of farmed crocodiles and ostriches. Rev Sci Tech 2002, 21,
                     265-276, doi:10.20506/rst.21.2.1334.
             14.     Horner, R.F. Poxvirus in farmed Nile crocodiles. Vet Rec 1988, 122, 459-462,
                     doi:10.1136/vr.122.19.459.
             15.     Gerdes, G.H. Morphology of poxviruses from reptiles. Vet Rec 1991, 128, 452,
                     doi:10.1136/vr.128.19.452.
             16.     Moore, R.L.; Isberg, S.R.; Shilton, C.M.; Milic, N.L. Impact of poxvirus lesions on saltwater
                     crocodile (Crocodylus porosus) skins. Vet Microbiol 2017, 211, 29-35,
                     doi:10.1016/j.vetmic.2017.09.019.

                                                                                                                 13
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022           doi:10.20944/preprints202205.0393.v1

             17.     Moss, B. Poxvirus entry and membrane fusion. Virology 2006, 344, 48-54,
                     doi:10.1016/j.virol.2005.09.037.
             18.     Bowie, A.G.; Unterholzner, L. Viral evasion and subversion of pattern-recognition
                     receptor signalling. Nat Rev Immunol 2008, 8, 911-922, doi:10.1038/nri2436.
             19.     Yu, H.; Bruneau, R.C.; Brennan, G.; Rothenburg, S. Battle Royale: Innate Recognition of
                     Poxviruses and Viral Immune Evasion. Biomedicines 2021, 9,
                     doi:10.3390/biomedicines9070765.
             20.     Meurs, E.; Chong, K.; Galabru, J.; Thomas, N.S.; Kerr, I.M.; Williams, B.R.; Hovanessian,
                     A.G. Molecular cloning and characterization of the human double-stranded RNA-
                     activated protein kinase induced by interferon. Cell 1990, 62, 379-390,
                     doi:10.1016/0092-8674(90)90374-n.
             21.     Beattie, E.; Tartaglia, J.; Paoletti, E. Vaccinia virus-encoded eIF-2 alpha homolog
                     abrogates the antiviral effect of interferon. Virology 1991, 183, 419-422,
                     doi:10.1016/0042-6822(91)90158-8.
             22.     Chang, H.W.; Watson, J.C.; Jacobs, B.L. The E3L gene of vaccinia virus encodes an
                     inhibitor of the interferon-induced, double-stranded RNA-dependent protein kinase.
                     Proc Natl Acad Sci U S A 1992, 89, 4825-4829, doi:10.1073/pnas.89.11.4825.
             23.     Davies, M.V.; Chang, H.W.; Jacobs, B.L.; Kaufman, R.J. The E3L and K3L vaccinia virus
                     gene products stimulate translation through inhibition of the double-stranded RNA-
                     dependent protein kinase by different mechanisms. J Virol 1993, 67, 1688-1692,
                     doi:10.1128/JVI.67.3.1688-1692.1993.
             24.     Langland, J.O.; Cameron, J.M.; Heck, M.C.; Jancovich, J.K.; Jacobs, B.L. Inhibition of PKR
                     by RNA and DNA viruses. Virus Res 2006, 119, 100-110,
                     doi:10.1016/j.virusres.2005.10.014.
             25.     Kerr, I.M.; Brown, R.E.; Hovanessian, A.G. Nature of inhibitor of cell-free protein
                     synthesis formed in response to interferon and double-stranded RNA. Nature 1977, 268,
                     540-542, doi:10.1038/268540a0.
             26.     Roberts, W.K.; Hovanessian, A.; Brown, R.E.; Clemens, M.J.; Kerr, I.M. Interferon-
                     mediated protein kinase and low-molecular-weight inhibitor of protein synthesis.
                     Nature 1976, 264, 477-480, doi:10.1038/264477a0.
             27.     Galabru, J.; Hovanessian, A. Autophosphorylation of the protein kinase dependent on
                     double-stranded RNA. J Biol Chem 1987, 262, 15538-15544.
             28.     Wu, S.; Kaufman, R.J. A model for the double-stranded RNA (dsRNA)-dependent
                     dimerization and activation of the dsRNA-activated protein kinase PKR. J Biol Chem
                     1997, 272, 1291-1296, doi:10.1074/jbc.272.2.1291.
             29.     Dar, A.C.; Dever, T.E.; Sicheri, F. Higher-order substrate recognition of eIF2alpha by the
                     RNA-dependent protein kinase PKR. Cell 2005, 122, 887-900,
                     doi:10.1016/j.cell.2005.06.044.
             30.     Dey, M.; Cao, C.; Dar, A.C.; Tamura, T.; Ozato, K.; Sicheri, F.; Dever, T.E. Mechanistic link
                     between PKR dimerization, autophosphorylation, and eIF2alpha substrate recognition.
                     Cell 2005, 122, 901-913, doi:10.1016/j.cell.2005.06.041.
             31.     Garcia, M.A.; Gil, J.; Ventoso, I.; Guerra, S.; Domingo, E.; Rivas, C.; Esteban, M. Impact of
                     protein kinase PKR in cell biology: from antiviral to antiproliferative action. Microbiol
                     Mol Biol Rev 2006, 70, 1032-1060, doi:10.1128/MMBR.00027-06.

                                                                                                                14
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022          doi:10.20944/preprints202205.0393.v1

             32.     Dey, M.; Mann, B.R.; Anshu, A.; Mannan, M.A. Activation of protein kinase PKR requires
                     dimerization-induced cis-phosphorylation within the activation loop. J Biol Chem 2014,
                     289, 5747-5757, doi:10.1074/jbc.M113.527796.
             33.     Guerra, S.; Lopez-Fernandez, L.A.; Conde, R.; Pascual-Montano, A.; Harshman, K.;
                     Esteban, M. Microarray analysis reveals characteristic changes of host cell gene
                     expression in response to attenuated modified vaccinia virus Ankara infection of human
                     HeLa cells. J Virol 2004, 78, 5820-5834, doi:10.1128/JVI.78.11.5820-5834.2004.
             34.     Lee, S.B.; Esteban, M. The interferon-induced double-stranded RNA-activated protein
                     kinase induces apoptosis. Virology 1994, 199, 491-496, doi:10.1006/viro.1994.1151.
             35.     Zhang, P.; Samuel, C.E. Protein kinase PKR plays a stimulus- and virus-dependent role in
                     apoptotic death and virus multiplication in human cells. J Virol 2007, 81, 8192-8200,
                     doi:10.1128/JVI.00426-07.
             36.     Yu, H.; Peng, C.; Zhang, C.; Stoian, A.M.M.; Tazi, L.; Brennan, G.; Rothenburg, S.
                     Maladaptation after a virus host switch leads to increased activation of the pro-
                     inflammatory NF-kappaB pathway. Proc Natl Acad Sci U S A 2022, 119, e2115354119,
                     doi:10.1073/pnas.2115354119.
             37.     Bratke, K.A.; McLysaght, A.; Rothenburg, S. A survey of host range genes in poxvirus
                     genomes. Infect Genet Evol 2013, 14, 406-425, doi:10.1016/j.meegid.2012.12.002.
             38.     Senkevich, T.G.; Yutin, N.; Wolf, Y.I.; Koonin, E.V.; Moss, B. Ancient Gene Capture and
                     Recent Gene Loss Shape the Evolution of Orthopoxvirus-Host Interaction Genes. mBio
                     2021, 12, e0149521, doi:10.1128/mBio.01495-21.
             39.     Watson, J.C.; Chang, H.W.; Jacobs, B.L. Characterization of a vaccinia virus-encoded
                     double-stranded RNA-binding protein that may be involved in inhibition of the double-
                     stranded RNA-dependent protein kinase. Virology 1991, 185, 206-216,
                     doi:10.1016/0042-6822(91)90768-7.
             40.     Chang, H.W.; Jacobs, B.L. Identification of a conserved motif that is necessary for
                     binding of the vaccinia virus E3L gene products to double-stranded RNA. Virology 1993,
                     194, 537-547, doi:10.1006/viro.1993.1292.
             41.     Romano, P.R.; Zhang, F.; Tan, S.L.; Garcia-Barrio, M.T.; Katze, M.G.; Dever, T.E.;
                     Hinnebusch, A.G. Inhibition of double-stranded RNA-dependent protein kinase PKR by
                     vaccinia virus E3: role of complex formation and the E3 N-terminal domain. Mol Cell Biol
                     1998, 18, 7304-7316.
             42.     Dar, A.C.; Sicheri, F. X-ray crystal structure and functional analysis of vaccinia virus K3L
                     reveals molecular determinants for PKR subversion and substrate recognition. Mol Cell
                     2002, 10, 295-305.
             43.     Little, N.S.; Quon, T.; Upton, C. Prediction of a novel RNA binding domain in crocodilepox
                     Zimbabwe Gene 157. Microb Inform Exp 2011, 1, 12, doi:10.1186/2042-5783-1-12.
             44.     Rahman, M.M.; Liu, J.; Chan, W.M.; Rothenburg, S.; McFadden, G. Myxoma virus protein
                     M029 is a dual function immunomodulator that inhibits PKR and also conscripts
                     RHA/DHX9 to promote expanded host tropism and viral replication. PLoS Pathog 2013,
                     9, e1003465, doi:10.1371/journal.ppat.1003465.
             45.     Rothenburg, S.; Seo, E.J.; Gibbs, J.S.; Dever, T.E.; Dittmar, K. Rapid evolution of protein
                     kinase PKR alters sensitivity to viral inhibitors. Nat Struct Mol Biol 2009, 16, 63-70,
                     doi:10.1038/nsmb.1529.

                                                                                                               15
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022           doi:10.20944/preprints202205.0393.v1

             46.     Peng, C.; Haller, S.L.; Rahman, M.M.; McFadden, G.; Rothenburg, S. Myxoma virus M156
                     is a specific inhibitor of rabbit PKR but contains a loss-of-function mutation in Australian
                     virus isolates. Proc Natl Acad Sci U S A 2016, 113, 3855-3860,
                     doi:10.1073/pnas.1515613113.
             47.     Vipat, S.; Brennan, G.; Park, C.; Haller, S.L.; Rothenburg, S. Rapid, Seamless Generation
                     of Recombinant Poxviruses using Host Range and Visual Selection. J Vis Exp 2020,
                     doi:10.3791/61049.
             48.     Dueck, K.J.; Hu, Y.S.; Chen, P.; Deschambault, Y.; Lee, J.; Varga, J.; Cao, J. Mutational
                     analysis of vaccinia virus E3 protein: the biological functions do not correlate with its
                     biochemical capacity to bind double-stranded RNA. J Virol 2015, 89, 5382-5394,
                     doi:10.1128/JVI.03288-14.
             49.     Edgar, R.C. MUSCLE: multiple sequence alignment with high accuracy and high
                     throughput. Nucleic Acids Res 2004, 32, 1792-1797, doi:10.1093/nar/gkh340.
             50.     Guindon, S.; Dufayard, J.F.; Lefort, V.; Anisimova, M.; Hordijk, W.; Gascuel, O. New
                     algorithms and methods to estimate maximum-likelihood phylogenies: assessing the
                     performance of PhyML 3.0. Syst Biol 2010, 59, 307-321, doi:10.1093/sysbio/syq010.
             51.     Schindelin, J.; Arganda-Carreras, I.; Frise, E.; Kaynig, V.; Longair, M.; Pietzsch, T.;
                     Preibisch, S.; Rueden, C.; Saalfeld, S.; Schmid, B.; et al. Fiji: an open-source platform for
                     biological-image analysis. Nat Methods 2012, 9, 676-682, doi:10.1038/nmeth.2019.
             52.     Bolte, S.; Cordelieres, F.P. A guided tour into subcellular colocalization analysis in light
                     microscopy. J Microsc 2006, 224, 213-232, doi:10.1111/j.1365-2818.2006.01706.x.
             53.     Manders, E.M.M.; Verbeek, F.J.; Aten, J.A. Measurement of co-localization of objects in
                     dual-colour confocal images. J Microsc 1993, 169, 375-382, doi:10.1111/j.1365-
                     2818.1993.tb03313.x.
             54.     Tian, B.; Mathews, M.B. Phylogenetics and functions of the double-stranded RNA-
                     binding motif: a genomic survey. Prog Nucleic Acid Res Mol Biol 2003, 74, 123-158,
                     doi:10.1016/s0079-6603(03)01012-2.
             55.     Ho, C.K.; Shuman, S. Mutational analysis of the vaccinia virus E3 protein defines amino
                     acid residues involved in E3 binding to double-stranded RNA. J Virol 1996, 70, 2611-
                     2614, doi:10.1128/JVI.70.4.2611-2614.1996.
             56.     Ramos, A.; Grunert, S.; Adams, J.; Micklem, D.R.; Proctor, M.R.; Freund, S.; Bycroft, M.;
                     St Johnston, D.; Varani, G. RNA recognition by a Staufen double-stranded RNA-binding
                     domain. EMBO J 2000, 19, 997-1009, doi:10.1093/emboj/19.5.997.
             57.     Nejepinska, J.; Malik, R.; Wagner, S.; Svoboda, P. Reporters transiently transfected into
                     mammalian cells are highly sensitive to translational repression induced by dsRNA
                     expression. PLoS One 2014, 9, e87517, doi:10.1371/journal.pone.0087517.
             58.     Elde, N.C.; Child, S.J.; Geballe, A.P.; Malik, H.S. Protein kinase R reveals an evolutionary
                     model for defeating viral mimicry. Nature 2009, 457, 485-489,
                     doi:10.1038/nature07529.
             59.     Park, C.; Peng, C.; Brennan, G.; Rothenburg, S. Species-specific inhibition of antiviral
                     protein kinase R by capripoxviruses and vaccinia virus. Ann N Y Acad Sci 2019, 1438, 18-
                     29, doi:10.1111/nyas.14000.
             60.     Park, C.; Peng, C.; Rahman, M.J.; Haller, S.L.; Tazi, L.; Brennan, G.; Rothenburg, S.
                     Orthopoxvirus K3 orthologs show virus- and host-specific inhibition of the antiviral

                                                                                                                16
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022             doi:10.20944/preprints202205.0393.v1

                     protein kinase PKR. PLoS Pathog 2021, 17, e1009183,
                     doi:10.1371/journal.ppat.1009183.
             61.     Mukaka, M.M. Statistics corner: A guide to appropriate use of correlation coefficient in
                     medical research. Malawi Med J 2012, 24, 69-71.
             62.     Sarker, S.; Isberg, S.R.; Moran, J.L.; Araujo, R.; Elliott, N.; Melville, L.; Beddoe, T.; Helbig,
                     K.J. Crocodilepox Virus Evolutionary Genomics Supports Observed Poxvirus Infection
                     Dynamics on Saltwater Crocodile (Crocodylus porosus). Viruses 2019, 11,
                     doi:10.3390/v11121116.
             63.     Odom, M.R.; Hendrickson, R.C.; Lefkowitz, E.J. Poxvirus protein evolution: family wide
                     assessment of possible horizontal gene transfer events. Virus Res 2009, 144, 233-249,
                     doi:10.1016/j.virusres.2009.05.006.
             64.     Senkevich, T.G.; Koonin, E.V.; Moss, B. Vaccinia virus F16 protein, a predicted
                     catalytically inactive member of the prokaryotic serine recombinase superfamily, is
                     targeted to nucleoli. Virology 2011, 417, 334-342, doi:10.1016/j.virol.2011.06.017.
             65.     Masliah, G.; Barraud, P.; Allain, F.H. RNA recognition by double-stranded RNA binding
                     domains: a matter of shape and sequence. Cell Mol Life Sci 2013, 70, 1875-1895,
                     doi:10.1007/s00018-012-1119-x.
             66.     Thakur, M.; Seo, E.J.; Dever, T.E. Variola virus E3L Zalpha domain, but not its Z-DNA
                     binding activity, is required for PKR inhibition. RNA 2014, 20, 214-227,
                     doi:10.1261/rna.042341.113.
             67.     Langland, J.O.; Jacobs, B.L. Inhibition of PKR by vaccinia virus: role of the N- and C-
                     terminal domains of E3L. Virology 2004, 324, 419-429, doi:10.1016/j.virol.2004.03.012.
             68.     Rojas, M.; Vasconcelos, G.; Dever, T.E. An eIF2alpha-binding motif in protein
                     phosphatase 1 subunit GADD34 and its viral orthologs is required to promote
                     dephosphorylation of eIF2alpha. Proc Natl Acad Sci U S A 2015, 112, E3466-3475,
                     doi:10.1073/pnas.1501557112.
             69.     Denzler, K.L.; Schriewer, J.; Parker, S.; Werner, C.; Hartzler, H.; Hembrador, E.; Huynh, T.;
                     Holechek, S.; Buller, R.M.; Jacobs, B.L. The attenuated NYCBH vaccinia virus deleted for
                     the immune evasion gene, E3L, completely protects mice against heterologous
                     challenge with ectromelia virus. Vaccine 2011, 29, 9691-9696,
                     doi:10.1016/j.vaccine.2011.09.108.
             70.     Denzler, K.L.; Rice, A.D.; MacNeill, A.L.; Fukushima, N.; Lindsey, S.F.; Wallace, G.;
                     Burrage, A.M.; Smith, A.J.; Manning, B.R.; Swetnam, D.M.; et al. The NYCBH vaccinia
                     virus deleted for the innate immune evasion gene, E3L, protects rabbits against lethal
                     challenge by rabbitpox virus. Vaccine 2011, 29, 7659-7669,
                     doi:10.1016/j.vaccine.2011.07.140.

                                                                                                                   17
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 30 May 2022                                                     doi:10.20944/preprints202205.0393.v1

             Figures

                                Rotavirus
                                 045        J NSP1 YP_010086031.1
                                Bat
                                 042 rotavirus NSP1 QRV11667.1

                              90 Porcine
                                   041     rotavirus H NGS-14 NSP1 BBC18340.1
                                     Rotavirus
                                      040       H SP-VC18 NSP1 QKY66963.1
                                               Bat
                                               0 4 3 rotavirus NSP1 QTE76090.1

                                                     Bat
                                                      0 4 4 rotavirus H NSP1 AWV67033.1

                                          Rotavirus
                                          038           I KE528/2012 NSP1-2 AKA63280.1
                                          Rotavirus
                                           039           I SD-F3 NSP1 ULF48556.1
                                                                                                         100       Nile
                                                                                                                    001 crocodilepox virus 157 YP_784347.1

                                                                                                                  Saltwater
                                                                                                                  002       crocodilepox virus 198 QGT47498
                                                97     Human
                                                        019       rotavirus C NSP3 AGO58054.2
                                                                   Human
                                                                    020       rotavirus C NSP3 AAX21224.1
                                                            76                Bovine
                                                                               022        rotavirus C NSP3 BCU08699.1
                                                                   74                            Rotavirus
                                                                                                  017           C NSP3 AXQ59463.1
                                                                          67         Porcine
                                                                                      021         rotavirus C NSP3 BCU08769.1
                                                                                        Porcine
                                                                                        018         rotavirus B NSP2 QBJ02236.1
                                                                                                      Beihai
                                                                                                       027       shrimp virus 2 gp2 YP_009333523.1
                                79                                 Drosophila
                                                                    026             C virus ORF1 AWI97853.1
                                                                     Wenzhou
                                                                     025            picorna-like virus 47 APG78496.1
                                                               Mimivirus
                                                                032           LCMiAC02 3'-5' exonuclease QBK89441.1
                                                                             Kadipiro
                                                                              024          virus VP8 APG79137.1
                                                                                            92                   Satyrvirus
                                                                                                                  035            sp. ribonuclease 3 AYV85794.1
                                     50
                                                                                                     71        Moumouvirus
                                                                                                                030                  maliensis RNAse QGR54010.1
                                                                                                                 Megavirus
                                                                                                                  031            courdo7 ribonuclease 3 AEX61722.1
                                                             74
                                                                                                                 Dishui
                                                                                                                  047     Lake phycodnavirus 3 ribonuclease III QIG59698.1
                                                                                                                                 Dishui
                                                                                                                                  037     Lake phycodnavirus 4 RNAse III QIG59958.1
                                                                                     53                            Micromonas
                                                                                                                    046               pusilla virus PL1 ribonuclease III AET43609.1
                                                                                                               87         Micromonas
                                                                                                                           036              pusilla virus SP1 RNAse III YP_009664928.1
                                                                                                                        Micromonas
                                                                                                                        049               pusilla virus PL1 ribonuclease III AET43609.1
                                                                                                            Ostreococcus
                                                                                                             048                   tauri virus RT-2011 OtV6_130 AFC35038.1
                                                                                                                  Ostreococcus
                                                                                                                   028                lucimarinus virus 1 OlV1_149c YP_004061782.1
                                                                                                                              Ostreococcus
                                                                                                                                029              tauri virus 1 OTV1_125 YP_003212948.1
                                                                      Bufonid
                                                                        016        herpesvirus 1 ORF25 YP_009552888.1
                                                      54                        Armadillidium
                                                                                 003                  vulgare iridescent virus Ribonuclease III YP_009046792.1
                                                                                  100 Iridovirus
                                                                                              004          Liz-CrIV 340R QEA08313.1
                                                                                             Invertebrate
                                                                                              006               iridescent virus 6 340R NP_149803.1
                                                         85                                Invertebrate
                                                                                            007               iridovirus 25 Ribonuclease III YP_009010659.1
                                                                  Anopheles
                                                                    005             minimus iridovirus Ribonuclease III YP_009021208.1
                                                                                    011        Hubei picorna-like virus 55 AWS06670.1
                                                                        87 012
                                                                            Changjiang crawfish virus 6 YP_009336630.1
                                                             75             Changjiang
                                                                             034               crawfish virus 6 APG78044.1
                                                                                        Hubei
                                                                                        008       picorna-like virus 56 YP_009336543.1
                                                                                 Sitobion miscanthi virus 1 polyprotein QCI31816.1
                                                                                010
                                      68                               80
                                                                                 Rosy
                                                                                 013     apple aphid virus polyprotein ABB89048.1
                                                                        57 Acyrthosiphon
                                                                                009                   pisum virus protein P1 QAA78869.1
                                                                         63 Acyrthosiphon
                                                                                033                   pisum virus P1 QAA78871.1
                                                     57                                                     Solenopsis
                                                                                                             015             invicta virus 7 polyprotein QBL75890.1
                                                                                                   Averyanov
                                                                                                    023             virus RNA-dependent RNA polymerase QIS87995.1
                                                                                                                        Beihai
                                                                                                                         014        anemone virus 1 BVE94_gp2 YP_009333203.1
                                                                         Pseudocowpox virus 020 AFA41730.1
                                                                 90 0 6 0
                                                                       50                Orf
                                                                                          053 virus OV20.0 CAA10953.1

                                                                                                     Seal
                                                                                                      058 parapoxvirus 013 YP_009389309.1

                                           51                                Swinepox
                                                                               054            virus 032 NP_570192.1
                                                                                        Myxoma
                                                                                         052          virus M156 NP_051743.1
                                                                       54               Tanapox
                                                                                         051           virus 34 ABQ43508.1
                                                           53                52                Deerpox
                                                                                                061         virus W-1170-84 042 ABI99027.1
                                                                                                      Goatpox
                                                                                                       055          virus Pellor 030 YP_001293225.1
                                                                                          96                  Cotia
                                                                                                               062     virus SPAn232 051 YP_005296239.1
                                                                      50                                        BeAn
                                                                                                                 056     58058 virus E3 YP_009329659.1
                                                                                 73                Yokapox
                                                                                                     057          virus 037 YP_004821390.1
                                                                                             83            Vaccinia
                                                                                                           050          virus Copenhagen E3 P21081.1
                                                                                                           Raccoonpox
                                                                                                            059                virus 054 YP_009143366.1

                                                     0.6

             Figure 1. Phylogenetic relationship between viral dsRNA-binding domains.
             A maximum phylogenetic tree of dsRBDs from indicated viral proteins was constructed from a
             multiple sequence alignment generated with MUSCLE using PhyML. Numbers next to branches
             indicate bootstrap values ≥ 50. Accession numbers of proteins are indicated.

                                                                                                                                                                                   18
You can also read